Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
FASEB J ; 37(10): e23160, 2023 10.
Article in English | MEDLINE | ID: mdl-37750502

ABSTRACT

Glioblastoma multiforme (GBM) is the most malignant brain tumor with rapid angiogenesis. How to inhibit GBM angiogenesis is a key problem to be solved. To explore the targets of inhibiting GBM angiogenesis, this study confirmed that the expression of circMTA1 (hsa_circ_0033614) was significantly upregulated in human brain microvascular endothelial cells exposed to glioma cell-conditioned medium (GECs). The expression of circMTA1 in the cytoplasm was significantly higher than that in the nucleus. Upregulated circMTA1 in GECs can promote cell proliferation, migration, and tube formation. Further exploration of the circularization mechanism of circMTA1 confirmed that KHDRBS1 protein can bind to the upstream and downstream flanking sequences of circMTA1 and promote circMTA1 biogenesis by coordinating Alu element pairing. KHDRBS1 upregulated the proliferation, migration, and tube formation of GECs by promoting the biogenesis of circMTA1. CircMTA1 can encode the protein MTA1-134aa by internal ribosome entry site sequence-mediated translation mechanism, and promote the proliferation, migration, and tube formation of GECs through the encoded MTA1-134aa. This study provides a new target for inhibiting angiogenesis in brain GBM and a new strategy for improving the therapeutic efficacy of GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Glioblastoma/genetics , Endothelial Cells , Alu Elements , Brain Neoplasms/genetics , Cell Cycle Proteins , DNA-Binding Proteins , RNA-Binding Proteins , Adaptor Proteins, Signal Transducing
2.
Neural Regen Res ; 17(1): 170-177, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34100453

ABSTRACT

Ghrelin is a neuropeptide that has various physiological functions and has been demonstrated to be neuroprotective in a number of neurological disease models. However, the underlying mechanisms of ghrelin in Parkinson's disease remain largely unexplored. The current study aimed to study the effects of ghrelin in a 6-hydroxydopamine (6-OHDA)-induced Parkinson's disease model and evaluate the potential underlying mechanisms. In the present study, we treated an SH-SY5Y cell model with 6-OHDA, and observed that pretreatment with different concentrations of ghrelin (1, 10, and 100 nM) for 30 minutes relieved the neurotoxic effects of 6-OHDA, as revealed by Cell Counting Kit-8 and Annexin V/propidium iodide (PI) apoptosis assays. Reverse transcription quantitative polymerase chain reaction and western blot assay results demonstrated that 6-OHDA treatment upregulated α-synuclein and lincRNA-p21 and downregulated TG-interacting factor 1 (TGIF1), which was predicted as a potential transcription regulator of the gene encoding α-synuclein (SNCA). Ghrelin pretreatment was able to reverse the trends caused by 6-OHDA. The Annexin V/PI apoptosis assay results revealed that inhibiting either α-synuclein or lincRNA-p21 expression with small interfering RNA (siRNA) relieved 6-OHDA-induced cell apoptosis. Furthermore, inhibiting lincRNA-p21 also partially upregulated TGIF1. By retrieving information from a bioinformatics database and performing both double luciferase and RNA immunoprecipitation assays, we found that lincRNA-p21 and TGIF1 were able to form a double-stranded RNA-binding protein Staufen homolog 1 (STAU1) binding site and further activate the STAU1-mediated mRNA decay pathway. In addition, TGIF1 was able to transcriptionally regulate α-synuclein expression by binding to the promoter of SNCA. The Annexin V/PI apoptosis assay results showed that either knockdown of TGIF1 or overexpression of lincRNA-p21 notably abolished the neuroprotective effects of ghrelin against 6-OHDA-induced neurotoxicity. Collectively, these findings suggest that ghrelin exerts neuroprotective effects against 6-OHDA-induced neurotoxicity via the lincRNA-p21/TGIF1/α-synuclein pathway.

3.
Front Psychol ; 12: 747719, 2021.
Article in English | MEDLINE | ID: mdl-35002844

ABSTRACT

To solve the problem that lack of interaction in online courses affects motivation and effectiveness of students' learning, smart interactive tools were introduced into the online Neurobiology course. This study aimed to evaluate the students' satisfaction with online teaching mode and assess the academically higher and lower performing students' learning effectiveness in the online course optimized with smart interactive tools compared to face-to-face learning. Descriptive statistics and independent t-tests were used to describe student samples and determine the differences in students' satisfaction and performance. Reflections of students' satisfaction revealed that about 65.8% were satisfied with the learning involvement and about 60.5% were satisfied with the class interaction. Almost two-thirds of the class agreed that the smart interactive tools applied in the online course could help them attain their learning goals better. Among all the smart interactive functions, the class quiz was the most effective one in helping students grasp the main points of the course. No significant differences were found between the two teaching modes in the overall and academically higher or lower performing students' final exam average scores. Compared to each band score of such two teaching modes, no one failed to pass the final exam in the online course, however, three lower-performing students who were taught in the traditional course failed. This study suggested that optimized online teaching with smart interactive tools could produce the same learning effectiveness for the academically lower-performing students as for the higher-performing students. Meanwhile, the instructors could know the learning status in which each student was and perform personalized guidance and improve exam passing rate accordingly.

4.
Oxid Med Cell Longev ; 2020: 8708236, 2020.
Article in English | MEDLINE | ID: mdl-32104542

ABSTRACT

Senescence of renal tubular epithelial cells plays an important role in diabetic nephropathy, but the mechanism is unknown. Metformin may alleviate diabetic nephropathy by reducing this senescence. This study is aimed at clarifying the effects and mechanism of metformin on the senescence of renal tubular epithelial cells in diabetic nephropathy. We found that metformin reduced the expression of senescence-associated gene P21 in high-glucose-induced (30 mmol/L) renal tubular epithelial cells and decreased the ß-galactosidase positive staining rate (decreased 16%, p < 0.01). Metformin was able to reduce senescence by upregulating the expression of RNA-binding protein MBNL1 and miR-130a-3p and reducing STAT3 expression. MBNL1 prolonged the half-life of miR-130a-3p, and miR-130a-3p could negatively regulate STAT3 by binding to its mRNA 3'UTR. In db/db diabetic mice, we found an enhanced senescence level combined with low expression of MBNL1 and miR-130a-3p and high expression of STAT3 compared with db/m control mice during nephropathy development. Meanwhile, metformin (200 mg/kg/day) could increase the expression of MBNL1 and miR-130a-3p and decreased STAT3 expression, thus reducing this senescence in db/db mice. Our results suggest that metformin reduces the senescence of renal tubular epithelial cells in diabetic nephropathy via the MBNL1/miR-130a-3p/STAT3 pathway, which provided new ideas for the therapy of this disease.


Subject(s)
Cellular Senescence/drug effects , DNA-Binding Proteins/metabolism , Diabetic Nephropathies/drug therapy , Metformin/therapeutic use , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Kidney Tubules/cytology , Mice , MicroRNAs/genetics , RNA-Binding Proteins/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects
5.
National Journal of Andrology ; (12): 110-117, 2019.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-816851

ABSTRACT

Objective@#To investigate the influence of the degrees of intravesical prostatic protrusion (IPP) on the recovery of urinary continence after radical prostatectomy.@*METHODS@#We retrospectively analyzed the clinical data on 212 patients diagnosed with prostate cancer by biopsy and treated by laparoscopic radical prostatectomy by the same surgeon. Based on the degrees of IPP measured by MRI, we divided the patients into an IPP ≤ 10 mm group (n = 146) and an IPP > 10 mm group (n = 66) and determined the factors influencing the recovery of urinary continence by univariate and multivariate logistic regression analyses.@*RESULTS@#At 1, 3, 6 and 12 months after surgery, the urinary continence rates of the patients were 32.5%, 50.5%, 82.1% and 91%, respectively. Univariate analysis indicated that the factors influencing the recovery of urinary continence included IPP, body mass index (BMI), bladder neck preservation (BNP), neurovascular bundle preservation (NVBP) and clinical tumor (T) stage at 3 months (P 10 mm (P 10 mm and BMI ≥ 25 kg/m2 are independent factors influencing the long-term recovery of urinary continence after radical prostatectomy.

6.
Cell Signal ; 30: 142-153, 2017 01.
Article in English | MEDLINE | ID: mdl-27693218

ABSTRACT

Malignant glioma is undoubtedly the most vascularized tumor of central nervous system. Angiogenesis, playing a predominant role in tumor progression, is widely considered as a key point of tumor treatment. The aim of this study was to investigate the potential effects of miR-383 on proliferation, migration, tube formation and angiogenesis of glioma-exposed endothelial cells (GECs) in vitro and to further elucidate its possible molecular mechanisms. The expression of miR-383 in GECs was significantly downregulated compared with that in normal endothelial cells (ECs). Overexpression of miR-383 dramatically inhibited the proliferation, migration, tube formation and spheroid-based angiogenesis of GECs in vitro. Dual-luciferase reporter results demonstrated vascular endothelial growth factor (VEGF) is a target gene of miR-383. Furthermore, overexpression or silencing of either miR-383 or VEGF was performed simultaneously to further clarify that miR-383 inhibited proliferation, migration and angiogenesis of GECs in vitro by targeting VEGF. Finally, VEGF/VEGFR2-mediated FAK and Src signaling pathways might contribute to anti-angiogenesis of GECs. In conclusion, our present study indicated that miR-383 inhibits proliferation, migration and angiogenesis of GECs in vitro via VEGF/VEGFR2-mediated FAK and Src signaling pathways, which would draw growing attention to miR-383c as a potential therapeutical target of glioma.


Subject(s)
Cell Movement/genetics , Endothelial Cells/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glioma/blood supply , Glioma/pathology , MicroRNAs/metabolism , Vascular Endothelial Growth Factor A/metabolism , src-Family Kinases/metabolism , Base Sequence , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioma/genetics , Humans , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Phosphorylation , Signal Transduction , Transfection , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Front Cell Neurosci ; 10: 235, 2016.
Article in English | MEDLINE | ID: mdl-27766072

ABSTRACT

Tumour suppressor candidate 7 (TUSC7) is a novel tumor suppressor gene generating long non-coding RNA (lncRNAs) in several types of human cancers. The expression and function of TUSC7 in human brain glioma has yet to be elucidated. In this study, TUSC7 was poorly expressed in tissues and cell lines of glioma, and the lower expression was correlated with glioma of the worse histological grade. Moreover, TUSC7 is a prognostic biomarker of glioma patients. Up-regulation of TUSC7 suppressed cellular proliferation and invasion of glioma cells, and accelerated cellular apoptosis. Bioinformatics analysis showed that TUSC7 specifically binds to miR-23b. MiR-23b was up-regulated in glioma and negatively correlated with the expression of TUSC7. The miR-23b expression was inhibited remarkably by the upregulation of TUSC7 and the reciprocal inhibition was determined between TUSC7 and miR-23b.RNA pull-down and luciferase reporter assays were used to validate the sequence-specific correlation between miR-23b and TUSC7. TUSC7 inhibited the proliferation, migration and invasion of glioma cells and promoted cellular apoptosis largely bypassing miR-23b. We conclude that the lncRNA TUSC7 acted as a tumor suppressor gene negatively regulated by miR-23b, suggesting a novel therapeutic strategy against gliomas.

8.
J Neurol Sci ; 369: 109-118, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27653874

ABSTRACT

NS1619, a calcium-activated potassium channel (Kca channel) activator, can selectively and time-dependently accelerate the formation of transport vesicles in both the brain tumor capillary endothelium and tumor cells within 15min of treatment and then increase the permeability of the blood-brain tumor barrier (BTB). However, the mechanism involved is still under investigation. Using a rat brain glioma (C6) model, the expression of caveolin-1, FoxO1 and p-FoxO1 protein were examined at different time points after intracarotid infusion of NS1619 at a dose of 30µg/kg/min. Internalization of Cholera toxin subunit (CTB) labeled fluorescently was monitored by flow cytometry. The expression of caveolin-1 and FoxO1 protein at tumor microvessels was enhanced and caveolae-mediated CTB endocytosis was increased by NS1619 infusion for 15min. Compared with the 15min group, the expression of caveolin-1 protein was significantly decreased and the level of phosphorylation of FoxO1 was significantly increased in the NS1619 2h group. In addition, inhibitors of reactive oxygen species (ROS) or PI3K or PKB significantly attenuated the level of FoxO1 phosphorylation and also increased the expression of caveolin-1 protein in Human Brain Microvascular Endothelial Cells (HBMECs) cocultured with human glioma cells (U87) 2h after NS1619 treatment. This led to the conclusion that NS1619-mediated transport vesicle increase is, at least partly, related to the ROS/PI3K/PKB/FoxO1 signaling pathway.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Caveolin 1/metabolism , Endothelial Cells/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Brain Neoplasms/pathology , Carotid Arteries/cytology , Caveolin 1/genetics , Cell Line, Tumor , Cholera Toxin/metabolism , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Glioma/pathology , Humans , Male , Morpholines/pharmacology , Rats , Rats, Wistar , Time Factors , Xenograft Model Antitumor Assays
9.
Med Sci Monit ; 22: 710-6, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26936749

ABSTRACT

BACKGROUND Human brain glioma is the most common endocranial tumor; its mortality and morbidity are very high. The objective of this study was to determine whether miR-338-3p can regulate malignant biological behaviors of glioma cells by targeted silencing of MACC1. MATERIAL AND METHODS The expression of miR-338-3p was detected by quantitative real-time PCR in brain glioma tissues and cell lines. Bioinformatics software was used to predict some potential target genes of miR-338-3p. Luciferase activities assay was used to verify the combination between target genes and miR-338-3p. And MACC1 protein expression was detected by Western blot. The apoptosis and proliferation ability were analyzed by MTT and flow cytometry assay. RESULTS Compared with normal brain tissues and cells, miR-338-3p in glioma tissues and cell lines was confirmed to be expressed at low levels, and down-regulation of miR-338-3p tended to be correlated with worse histological grade. Up-regulation of miR-338-3p promoted apoptosis and sharply inhibited cell proliferation ability of U251 and U87 cells. The luciferase activities assay, biotin-avidin pull-down assay, and western blot analysis verified that MACC1 was a specific target gene of miR-338-3p. Subsequent experiments found that up-regulation of MACC1 significantly inhibited the apoptosis and increased the cell proliferation ability of U251 and U87 cells. The regulation effects of miR-338-3p on malignant biological behaviors of glioma cells can be partly reversed by up-regulation of MACC1. CONCLUSIONS Down-regulation of miR-338-3p was an independent prognostic biomarker associated with poor prognosis in glioma patients; miR-338-3p acted as a tumor-suppressing gene whose silencing can inhibit malignant biological behaviors of glioma cells. MACC1 was a specific target gene of miR-338-3p, which regulates malignant biological behaviors of glioma cells partly through directly silencing MACC1 expression.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Genes, Neoplasm , Glioma/genetics , Glioma/pathology , MicroRNAs/metabolism , Transcription Factors/genetics , 3' Untranslated Regions/genetics , Base Sequence , Binding Sites , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators , Transcription Factors/metabolism , Transfection , Up-Regulation/genetics
10.
Front Mol Neurosci ; 9: 9, 2016.
Article in English | MEDLINE | ID: mdl-26903801

ABSTRACT

The primary goals of this study were to investigate the potential roles of miR-200b in regulating RMP7-induced increases in blood-tumor barrier (BTB) permeability and some of the possible molecular mechanisms associated with this effect. Microarray analysis revealed 34 significantly deregulated miRNAs including miR-200b in the BTB as induced by RMP7 and 8 significantly up-regulated miRNAs in the BTB by RMP7. RMP7 induced tight junction (TJ) opening of the BTB, thereby increasing BTB permeability. Associated with this effect of RMP7 was a decrease in miR-200b expression within the human cerebral microvascular endothelial cells line hCMEC/D3 (ECs) of the BTB. Overexpression of miR-200b inhibited endothelial leakage and restored normal transendothelial electric resistance values. A simultaneous shift in occludin and claudin-5 distributions from insoluble to soluble fractions were observed to be significantly reduced. In addition, overexpression of miR-200b inhibited the relocation of occludin and claudin-5 from cellular borders into the cytoplasm as well as the production of stress fiber formation in GECs (ECs with U87 glioma cells co-culturing) of the BTB. MiR-200b silencing produced opposite results as that obtained from that of the miR-200b overexpression group. Overexpression of miR-200b was also associated with a down-regulation in RhoA and ROCKII expression, concomitant with a decrease in BTB permeability. Again, results which were opposite to that obtained with the miR-200b silencing group. We further found that miR-200b regulated BTB permeability by directly targeting RhoA and ROCKII. Collectively, these results suggest that miR-200b's contribution to the RMP7-induced increase in BTB permeability was associated with stress fiber formation and TJ disassembly as achieved by directly targeting RhoA and ROCKII.

11.
CNS Neurosci Ther ; 22(6): 477-87, 2016 06.
Article in English | MEDLINE | ID: mdl-26861687

ABSTRACT

AIM: The nontoxic mutant of diphtheria toxin (DT) has been demonstrated to act as a receptor-specific carrier protein to delivery drug into brain. Recent research showed that the truncated "receptorless" DT was still capable of being internalized into cells. This study investigated the effects and potential mechanisms of DT(270-326) , a truncated "receptorless" DT, on the permeability of the blood-tumor barrier (BTB). METHODS: BTB and GECs were subjected to DT(270-326) treatment. HRP flux assays, immunofluorescent, co-immunoprecipitation, Western blot, CCK-8, and Flow cytometry analysis were used to evaluate the effects of DT(270-326) administration. RESULTS: Our results revealed that 5 µM of DT(270-326) significantly increased the permeability of BTBin vitro, which reached its peak at 6 h. The permeability was reduced by pretreatment with filipinIII. DT(270-326) co-localized and interacted with caveolin-1 via its caveolin-binding motif. The mRNA and protein expression levels of caveolin-1 were identical with the changes of BTB permeability. The upregulated expression of caveolin-1 was associated with Src kinase-dependent tyrosine phosphorylation of caveolin-1, which subsequently induced phosphorylation and inactivation of the transcription factor Egr-1. The combination of DT(270-326) with doxorubicin significantly enhanced the loss of cell viability and apoptosis of U87 glioma cells in contrast to doxorubicin alone. CONCLUSIONS: DT(270-326) might provide a novel strategy to increase the delivery of macromolecular therapeutic agents across the BTB.


Subject(s)
Capillary Permeability/drug effects , Diphtheria Toxin/metabolism , Transcytosis/drug effects , Up-Regulation/drug effects , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Capillary Permeability/genetics , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line, Transformed , Cell Line, Tumor , Diphtheria Toxin/chemistry , Diphtheria Toxin/genetics , Diphtheria Toxin/pharmacology , Drug Synergism , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Glioblastoma/ultrastructure , Horseradish Peroxidase/pharmacokinetics , Humans , Mutation/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors , Transcytosis/genetics , Up-Regulation/genetics
12.
Oncol Lett ; 11(1): 904-908, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26870304

ABSTRACT

In this study, we investigate whether miR-128 is capable of regulating the apoptosis and proliferation of human U251 glioma cells by downregulating RhoE. The expression of miR-128 was assessed by quantitative polymerase chain reaction in normal brain tissue and glioma samples. A significant downregulation of the expression of miR-128 was detected in glioma in contrast to normal brain tissue. Following the transfection of pre-miR-128 and anti-miR-128 into U251 cells, the high expression of miR-128 could inhibit proliferation and induce apoptosis in U251 cells, and those effects could be restored by miR-128 knockdown. To analyze the regulation mechanism of miR-128, TargetScan, miRanda and PicTar were used to ascertain whether RhoE was a potential target gene. Next, luciferase activity assay and western blot analysis confirmed that RhoE was a direct and specific target gene of miR-128. The advanced effects of pre-miR-128 on the apoptosis and proliferation of U251 cells were reversed by the upregulation of RhoE expression. In summary, aberrantly expressed miR-128 regulates apoptosis and proliferation in human glioma U251 cells partly by directly targeting RhoE. This finding may offer a new potential therapeutic strategy for the treatment of glioma.

13.
Cancer Chemother Pharmacol ; 77(3): 507-13, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26781446

ABSTRACT

PURPOSE: In this study, whether HOTAIR is a prognostic biomarker will be detected, and its regulative effects of chemosensitivity to doxorubicin in TCC cells will be examined. METHODS: The expression of HOTAIR was detected by quantitative real-time PCR. Overall survival rate was calculated by Kaplan-Meier method with the log-rank test for comparisons. MTT assay was used to detect cell proliferation ability and chemosensitivity. Dual-color flow cytometric method was used to detect cell apoptosis. RESULTS: HOTAIR was up-regulated in bladder transitional cell carcinoma (TCC) tissues and cell lines compared with normal bladder transitional cell (NBTC) tissues and bladder epithelial immortalized SV-HUC-1 cells, and its expression level had positive correlation with histological grades of TCC. Moreover, HOTAIR was an independent prognostic biomarker of overall survival for TCC patients. The expression and silence vector for HOTAIR were transfected into T24 and J82 cells to up-regulate and silence the HOTAIR expression, respectively. In T24 and J82 cells, HOTAIR over-expression promoted cell proliferation and inhibited chemosensitivity to doxorubicin and cell apoptosis induced by doxorubicin; silence of HOTAIR showed opposite regulative effects. CONCLUSIONS: In summary, lncRNA HOTAIR was an independent prognostic biomarker of overall survival in TCC patients and could regulate chemosensitivity to doxorubicin of human TCC cells. HOTAIR might provide a new potential therapeutic target and stratagem for TCC.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Carcinoma, Transitional Cell/drug therapy , Doxorubicin/pharmacology , RNA, Long Noncoding/genetics , Urinary Bladder Neoplasms/drug therapy , Aged , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/pathology , Cell Proliferation/drug effects , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/genetics , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Survival Rate , Up-Regulation/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
14.
J Mol Neurosci ; 58(2): 153-61, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26358039

ABSTRACT

Previous studies have demonstrated that low-dose endothelial monocyte-activating polypeptide-II (EMAP-II) induces blood-tumor barrier (BTB) hyperpermeability via both paracellular and transcellular pathways. In a recent study, we revealed that cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)-dependent signaling pathway is involved in EMAP-II-induced BTB hyperpermeability. This study further investigated the exact mechanisms through which the cAMP/PKA-dependent signaling pathway affects EMAP-II-induced BTB hyperpermeability. In an in vitro BTB model, low-dose EMAP-II (0.05 nM) induced a significant decrease in Rac1 activity in rat brain microvascular endothelial cells (RBMECs). Pretreatment with forskolin to elevate intracellular cAMP concentration completely blocked EMAP-II-induced inactivation of Rac1. Besides, pretreatment with 6Bnz-cAMP to activate PKA partially attenuated EMAP-II-induced Rac1 inactivation. Moreover, 6Bnz-cAMP pretreatment significantly diminished EMAP-II-induced changes in BTB permeability, myosin light chain (MLC) phosphorylation, expression and distribution of ZO-1, and actin cytoskeleton arrangement in RBMECs. These effects of 6Bnz-cAMP were completely blocked in the presence of NSC-23766 (the specific inhibitor of Rac1). In conclusion, this study demonstrates that low-dose EMAP-II induces BTB hyperpermeability via the cAMP/PKA/Rac1 signaling pathway.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Capillary Permeability , Cyclic AMP/metabolism , Cytokines/metabolism , Endothelium, Vascular/metabolism , Glioma/metabolism , Neoplasm Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Rats , Rats, Wistar , Second Messenger Systems , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism
15.
J Mol Neurosci ; 59(2): 193-202, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26521255

ABSTRACT

Previous studies have demonstrated that low-dose endothelial monocyte-activating polypeptide-II (EMAP-II) can increase blood-tumor barrier (BTB) permeability via both paracellular and transcellular pathways. In addition, we revealed that the RhoA/Rho kinase (ROCK) signaling pathway is involved in EMAP-II-induced BTB opening. This study further investigated the exact mechanisms by which the RhoA/ROCK signaling pathway affects EMAP-II-induced BTB hyperpermeability. In an in vitro BTB model, low-dose EMAP-II significantly activated phosphatidylinositol-3-kinase (PI3K) in rat brain microvascular endothelial cells (RBMECs) at 0.75 h. Pretreatment with RhoA inhibitor C3 exoenzyme or ROCK inhibitor Y-27632 completely blocked EMAP-II-induced activation of PI3K. PKC-α/ß inhibitor GÖ6976 pretreatment caused no change in EMAP-II-induced activation of PI3K. Besides, pretreatment with LY294002, a specific inhibitor of PI3K, did not affect EMAP-II-induced activation of PKC-α/ß. Furthermore, LY294002 pretreatment significantly diminished EMAP-II-induced changes in BTB permeability, phosphorylation of myosin light chain and cofilin, expression and distribution of tight junction-associated protein ZO-1, and actin cytoskeleton arrangement in RBMECs. In summary, this study demonstrates that low-dose EMAP-II can increase BTB permeability by activating the RhoA/ROCK/PI3K signaling pathway.


Subject(s)
Capillary Permeability/drug effects , Cytokines/pharmacology , Endothelium, Vascular/metabolism , Neoplasm Proteins/pharmacology , Neovascularization, Pathologic/metabolism , RNA-Binding Proteins/pharmacology , Signal Transduction , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Endothelium, Vascular/cytology , Myosin Light Chains/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Zonula Occludens-1 Protein/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
16.
National Journal of Andrology ; (12): 116-121, 2016.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-304741

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effects of the quinoline derivative PQ1 combined with cisplatin on the proliferation and gap junction communication of prostate cancer PC3 cells.</p><p><b>METHODS</b>We cultured in vitro prostate cancer PC3 cells and divided them into DMSO blank control, cisplatin control, and cisplatin (10 mg/ml) plus PQ1 (1, 2, 5, 10, and 15 μmol/L) groups. We measured the proliferation of the prostate cancer PC3 cells, determined the expressions of the connexin 43 (Cx43) mRNA and protein by RT-PCR and Western blot, and compared the indexes among different groups.</p><p><b>RESULTS</b>Cisplatin combined with PQl at 1 - 10 μmol/L significantly inhibited the proliferation of the PC3 cells and the inhibition rate rose in a concentration- and time-dependent manner, from (48.72 ± 0.98)% vs (50.33 ± 0.62)% at 0 μmol/L to (77.38 ± 1.12)% vs (83.50 ± 1.05)% at 15 μmol/L at 24 and 48 hours (P < 0.05). Compared with the cisplatin control, cisplatin combined with PQ1 at 1, 2, 5, 10, and 15 μmol/L increased the expression of Cx43 mRNA from 0.379 ± 0.113 to 0.669 ± 0.031, 0.831 ± 0. 127, 0.769 ± 0.100, 0.532 ± 0.086, and 0.475 ± 0.134, respectively (P < 0.05), and cisplatin combined with PQ1 at 1, 2, 5, and 10 μmol/L elevated that of Cx43 protein from 0.138 ± 0.146 to 0.263 ± 0.111, 0.306 ± 0.152, 0.415 ± 0.280, and 0.643 ± 0.310, respectively (P < 0.05).</p><p><b>CONCLUSION</b>The quinoline derivative PQ1 can promote the gap junction communication of prostate cancer PC3 cells and enhance the killing effect of cisplatin on PC3 cells by upregulating the expressions of Cx43 mRNA and protein.</p>


Subject(s)
Humans , Male , Aminoquinolines , Pharmacology , Antineoplastic Combined Chemotherapy Protocols , Pharmacology , Cell Line, Tumor , Cell Proliferation , Cisplatin , Pharmacology , Connexin 43 , Genetics , Metabolism , Dose-Response Relationship, Drug , Gap Junctions , Physiology , Prostatic Neoplasms , Metabolism , Pathology , RNA, Messenger , Metabolism , Time Factors
17.
National Journal of Andrology ; (12): 415-419, 2016.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-262336

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the factors upgrading the International Society of Urological Pathology (ISUP) Gleason score using the specimens from preoperative prostatic biopsy and radical prostatectomy.</p><p><b>METHODS</b>A total of 164 patients diagnosed with prostate cancer by biopsy underwent radical prostatectomy. We retrospectively analyzed their age, prostate volume, preoperative PSA level, PSA density (PSAD) , the time interval between biopsy and surgery, the number of positive punctures, positive surgical margin, seminal vesicle invasion, lymphatic invasion, and Gleason scores from biopsy and prostatectomy. We also determined the predictors of Gleason score upgrading by logistic regression analysis.</p><p><b>RESULTS</b>Of the 164 cases analyzed, 95 (57.93% ) showed a consistency between the Gleason score of preoperative prostatic biopsy and that after radical prostatectomy, 55 (33.54% ) increased and 14 (8.52%) decreased after prostatectomy as compared with preoperative biopsy. The prostate volume (P < 0.01) and biopsy score (P < 0.05) were independent predictors of Gleason score upgrading. The risk of Gleason score upgrading was 27 times higher in the patients with the prostate volume ≤ 25 ml and 9 times higher in the 25-40 ml group than in the > 60 ml group (P < 0.05).</p><p><b>CONCLUSION</b>Low Gleason score of biopsy (≤ 6) and small prostate volume (≤ 40 ml) may be the predictors of Gleason score upgrading after radical prostatectomy.</p>


Subject(s)
Humans , Male , Biopsy , Neoplasm Grading , Organ Size , Prostate-Specific Antigen , Blood , Prostatectomy , Prostatic Neoplasms , Classification , General Surgery , Retrospective Studies , Risk Factors
18.
Biochem Biophys Res Commun ; 468(1-2): 105-12, 2015.
Article in English | MEDLINE | ID: mdl-26541455

ABSTRACT

Therapeutic applications of microRNAs (miRNAs) in chemotherapy were confirmed to be valuable, but there is rare to identify their specific roles and functions in shikonin treatment toward tumors. Here, for the first time, we reported that miR-143 played a critical role in the antitumor activity of shikonin in glioblastoma stem cells (GSCs). The results showed that the expression of miR-143 was downregulated in shikonin treated GSCs within 24 h. MiR-143 overexpression significantly enhanced the inhibitory effect of shikonin toward GSCs on cell viability. Besides, miR-143 overexpression caused a significant increase in the apoptotic fraction and made apoptosis occur earlier. Further investigation identified that BAG3, an apoptotic regulator, was a functional target of miR-143 in shikonin treated GSCs. The expression of BAG3 was upregulated in shikonin treated GSCs within 24 h. MiR-143 overexpression significantly reversed the high expression of BAG3 in shikonin treated GSCs. Moreover, it was confirmed that the enhanced cytotoxicity of shikonin by miR-143 overexpression was reversed by BAG3 overexpression both in vitro and in vivo, suggesting that the enhanced tumor suppressive effects by miR-143 overexpression was at least partly through the regulation of BAG3. Taken together, for the first time, our results demonstrate that miR-143 could enhance the antitumor activity of shikonin toward GSCs through reducing BAG3 expression, which may provide a novel therapeutic strategy for enhancing the treatment efficacy of shikonin toward GSCs.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis Regulatory Proteins/genetics , Drugs, Chinese Herbal/therapeutic use , Glioblastoma/drug therapy , MicroRNAs/genetics , Naphthoquinones/therapeutic use , Neoplastic Stem Cells/drug effects , Animals , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured , Up-Regulation/drug effects
19.
Int J Mol Sci ; 16(10): 23823-48, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26473829

ABSTRACT

Shikonin is an anthraquinone derivative extracted from the root of lithospermum. Shikonin is traditionally used in the treatment of inflammatory and infectious diseases such as hepatitis. Shikonin also inhibits proliferation and induces apoptosis in various tumors. However, the effect of shikonin on gliomas has not been fully elucidated. In the present study, we aimed to investigate the effects of shikonin on the migration and invasion of human glioblastoma cells as well as the underlying mechanisms. U87 and U251 human glioblastoma cells were treated with shikonin at 2.5, 5, and 7.5 µmol/L and cell viability, migration and invasiveness were assessed with CCK8, scratch wound healing, in vitro Transwell migration, and invasion assays. The expression and activity of matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9) and the expression of phosphorylated ß-catenin (p-ß-catenin) and phosphorylated PI3K/Akt were also checked. Results showed that shikonin significantly inhibited the cell proliferation, migration, invasion, and expression of MMP-2 and MMP-9 in U87 and U251 cells. The expression of p-ß-catenin showed contrary trends in two cell lines. It was significantly inhibited in U87 cells and promoted in U251 cells. Results in this work indicated that shikonin displayed an inhibitory effect on the migration and invasion of glioma cells by inhibiting the expression and activity of MMP-2 and -9. In addition, shikonin also inhibited the expression of p-PI3K and p-Akt to attenuate cell migration and invasion and MMP-2 and MMP-9 expression in both cell lines, which could be reversed by the PI3K/Akt pathway agonist, insulin-like growth factor-1 (IGF-1).


Subject(s)
Cell Movement/drug effects , Glioblastoma/pathology , Naphthoquinones/pharmacology , Neoplasm Invasiveness/pathology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drugs, Chinese Herbal/pharmacology , Humans , Insulin-Like Growth Factor I , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/metabolism , Medicine, Chinese Traditional , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/drug effects , beta Catenin/genetics , beta Catenin/metabolism
20.
J Neurosci Res ; 93(12): 1891-902, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26356851

ABSTRACT

miR-18a represses angiogenesis and tumor evasion by weakening vascular endothelial growth factor and transforming growth factor-ß signaling to prolong the survival of glioma patients, although it is thought to be an oncogene. This study investigates the potential effects of miR-18a on the permeability of the blood-tumor barrier (BTB) and its possible molecular mechanisms. An in vitro BTB model was successfully established. The endogenous expression of miR-18a in glioma vascular endothelial cells (GECs) was significantly lower than that in normal vascular ECs, and the overexpression of miR-18a significantly increased the permeability of the BTB as well as downregulating the mRNA and protein expressions of tight junction-related proteins zonula occluden-1 (ZO-1), claudin-5, and occludin in GECs. Dual luciferase reporter assays revealed that miR-18a bound to the 3'-untranslated region (3'UTR) of myocyte enhancer factor 2D (MEF2D). The overexpression of both miR-18a and MEF2D with the 3'UTR significantly weakened the effect caused by miR-18a of decreasing the mRNA and protein expressions of ZO-1, claudin-5 and occludin and of increasing the permeability of the BTB. Chromatin immunoprecipitation showed that MEF2D could directly bind to KLF4 promoter. This study shows that miR-18a targets and negatively regulates MEF2D, which further regulates tight junction-related proteins ZO-1, claudin-5, and occludin through transactivation of KLF4 and, finally, changes the permeability of the BTB. MiR-18a should garner growing attention because it might serve as a potential target in opening the BTB and providing a new strategy for the treatment of gliomas.


Subject(s)
Down-Regulation/physiology , Epithelial Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , MEF2 Transcription Factors/metabolism , MicroRNAs/metabolism , Zonula Occludens Proteins/metabolism , Blood-Brain Barrier/cytology , Capillary Permeability/physiology , Cell Line, Transformed , Chromatin Immunoprecipitation , Claudin-5/metabolism , Glioma/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Horseradish Peroxidase/metabolism , Humans , Kruppel-Like Factor 4 , MicroRNAs/genetics , Occludin/metabolism , Permeability , RNA, Messenger/metabolism , Transfection , Zonula Occludens Proteins/genetics , Zonula Occludens-1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...